1
|
Ali M, Huarte OU, Heurtaux T, Garcia P, Rodriguez BP, Grzyb K, Halder R, Skupin A, Buttini M, Glaab E. Single-Cell Transcriptional Profiling and Gene Regulatory Network Modeling in Tg2576 Mice Reveal Gender-Dependent Molecular Features Preceding Alzheimer-Like Pathologies. Mol Neurobiol 2024; 61:541-566. [PMID: 35980567 PMCID: PMC10861719 DOI: 10.1007/s12035-022-02985-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 07/29/2022] [Indexed: 11/26/2022]
Abstract
Alzheimer's disease (AD) onset and progression is influenced by a complex interplay of several environmental and genetic factors, one of them gender. Pronounced gender differences have been observed both in the relative risk of developing AD and in clinical disease manifestations. A molecular level understanding of these gender disparities is still missing, but could provide important clues on cellular mechanisms modulating the disease and reveal new targets for gender-oriented disease-modifying precision therapies. We therefore present here a comprehensive single-cell analysis of disease-associated molecular gender differences in transcriptomics data from the neocortex, one of the brain regions most susceptible to AD, in one of the most widely used AD mouse models, the Tg2576 model. Cortical areas are also most commonly used in studies of post-mortem AD brains. To identify disease-linked molecular processes that occur before the onset of detectable neuropathology, we focused our analyses on an age with no detectable plaques and microgliosis. Cell-type specific alterations were investigated at the level of individual genes, pathways, and gene regulatory networks. The number of differentially expressed genes (DEGs) was not large enough to build context-specific gene regulatory networks for each individual cell type, and thus, we focused on the study of cell types with dominant changes and included analyses of changes across the combination of cell types. We observed significant disease-associated gender differences in cellular processes related to synapse organization and reactive oxygen species metabolism, and identified a limited set of transcription factors, including Egr1 and Klf6, as key regulators of many of the disease-associated and gender-dependent gene expression changes in the model. Overall, our analyses revealed significant cell-type specific gene expression changes in individual genes, pathways and sub-networks, including gender-specific and gender-dimorphic changes in both upstream transcription factors and their downstream targets, in the Tg2576 AD model before the onset of overt disease. This opens a window into molecular events that could determine gender-susceptibility to AD, and uncovers tractable target candidates for potential gender-specific precision medicine for AD.
Collapse
Affiliation(s)
- Muhammad Ali
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- School for Mental Health and Neuroscience (MHeNs), Department of Psychiatry and Neuropsychology, Maastricht University, 6200, Maastricht, the Netherlands
| | - Oihane Uriarte Huarte
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Tony Heurtaux
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- Department of Life Sciences and Medicine (DLSM), University of Luxembourg, L‑4362, Esch-Sur-Alzette, Luxembourg
| | - Pierre Garcia
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Beatriz Pardo Rodriguez
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
- University of the Basque Country, Cell Biology and Histology Department, 48940, Leioa, Vizcaya, Basque Country, Spain
| | - Kamil Grzyb
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Rashi Halder
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
| | - Alexander Skupin
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Department of Physics and Materials Science, University of Luxembourg, 162a av. de la Faïencerie, 1511, Luxembourg, Luxembourg
- Department of Neuroscience, University of California San Diego, 9500 Gilman Dr, La Jolla, CA, 92093, USA
| | - Manuel Buttini
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg
- Luxembourg Center of Neuropathology (LCNP), L-3555, Dudelange, Luxembourg
| | - Enrico Glaab
- Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, 7 avenue des Hauts Fourneaux, L-4362, Esch-sur-Alzette, Luxembourg.
| |
Collapse
|
2
|
Garcia G, Bar‐Ziv R, Averbukh M, Dasgupta N, Dutta N, Zhang H, Fan W, Moaddeli D, Tsui CK, Castro Torres T, Alcala A, Moehle EA, Hoang S, Shalem O, Adams PD, Thorwald MA, Higuchi‐Sanabria R. Large-scale genetic screens identify BET-1 as a cytoskeleton regulator promoting actin function and life span. Aging Cell 2023; 22:e13742. [PMID: 36404134 PMCID: PMC9835578 DOI: 10.1111/acel.13742] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 10/17/2022] [Accepted: 11/01/2022] [Indexed: 11/22/2022] Open
Abstract
The actin cytoskeleton is a three-dimensional scaffold of proteins that is a regulatory, energyconsuming network with dynamic properties to shape the structure and function of the cell. Proper actin function is required for many cellular pathways, including cell division, autophagy, chaperone function, endocytosis, and exocytosis. Deterioration of these processes manifests during aging and exposure to stress, which is in part due to the breakdown of the actin cytoskeleton. However, the regulatory mechanisms involved in preservation of cytoskeletal form and function are not well-understood. Here, we performed a multipronged, cross-organismal screen combining a whole-genome CRISPR-Cas9 screen in human fibroblasts with in vivo Caenorhabditis elegans synthetic lethality screening. We identified the bromodomain protein, BET-1, as a key regulator of actin function and longevity. Overexpression of bet-1 preserves actin function at late age and promotes life span and healthspan in C. elegans. These beneficial effects are mediated through actin preservation by the transcriptional regulator function of BET-1. Together, our discovery assigns a key role for BET-1 in cytoskeletal health, highlighting regulatory cellular networks promoting cytoskeletal homeostasis.
Collapse
Affiliation(s)
- Gilberto Garcia
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Raz Bar‐Ziv
- Department of Molecular & Cellular Biology, Howard Hughes Medical InstituteThe University of California, BerkeleyBerkeleyCaliforniaUSA
| | - Maxim Averbukh
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Nirmalya Dasgupta
- Aging, Cancer and Immuno‐oncology ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCaliforniaUSA
| | - Naibedya Dutta
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Hanlin Zhang
- Department of Molecular & Cellular Biology, Howard Hughes Medical InstituteThe University of California, BerkeleyBerkeleyCaliforniaUSA
| | - Wudi Fan
- Department of Molecular & Cellular Biology, Howard Hughes Medical InstituteThe University of California, BerkeleyBerkeleyCaliforniaUSA
| | - Darius Moaddeli
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - C. Kimberly Tsui
- Department of Molecular & Cellular Biology, Howard Hughes Medical InstituteThe University of California, BerkeleyBerkeleyCaliforniaUSA
| | - Toni Castro Torres
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Athena Alcala
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Erica A. Moehle
- Department of Molecular & Cellular Biology, Howard Hughes Medical InstituteThe University of California, BerkeleyBerkeleyCaliforniaUSA
| | - Sally Hoang
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Ophir Shalem
- Department of Genetics, Perelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Peter D. Adams
- Aging, Cancer and Immuno‐oncology ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCaliforniaUSA
| | - Max A. Thorwald
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Ryo Higuchi‐Sanabria
- Leonard Davis School of GerontologyUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| |
Collapse
|
3
|
Hu YT, Chen XL, Zhang YN, McGurran H, Stormmesand J, Breeuwsma N, Sluiter A, Zhao J, Swaab D, Bao AM. Sex differences in hippocampal β-amyloid accumulation in the triple-transgenic mouse model of Alzheimer's disease and the potential role of local estrogens. Front Neurosci 2023; 17:1117584. [PMID: 36968493 PMCID: PMC10030503 DOI: 10.3389/fnins.2023.1117584] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 02/22/2023] [Indexed: 03/29/2023] Open
Abstract
Introduction Epidemiological studies show that women have a higher prevalence of Alzheimer's disease (AD) than men. Peripheral estrogen reduction during aging in women is proposed to play a key role in this sex-associated prevalence, however, the underlying mechanism remains elusive. We previously found that transcription factor early growth response-1 (EGR1) significantly regulates cholinergic function. EGR1 stimulates acetylcholinesterase (AChE) gene expression and is involved in AD pathogenesis. We aimed to investigate whether the triple-transgenic AD (3xTg-AD) mice harboring PS1 M146V , APP Swe , and Tau P301L show sex differences in β-amyloid (Aβ) and hyperphosphorylated tau (p-Tau), the two primary AD hallmarks, and how local 17β-estradiol (E2) may regulate the expression of EGR1 and AChE. Methods We first sacrificed male and female 3xTg-AD mice at 3-4, 7-8, and 11-12 months and measured the levels of Aβ, p-Tau, EGR1, and AChE in the hippocampal complex. Second, we infected SH-SY5Y cells with lentivirus containing the amyloid precursor protein construct C99, cultured with or without E2 administration we measured the levels of extracellular Aβ and intracellular EGR1 and AChE. Results Female 3xTg-AD mice had higher levels of Aβ compared to males, while no p-Tau was found in either group. In SH-SY5Y cells infected with lentivirus containing the amyloid precursor protein construct C99, we observed significantly increased extracellular Aβ and decreased expression of intracellular EGR1 and AChE. By adding E2 to the culture medium, extracellular Aβ(l-42) was significantly decreased while intracellular EGR1 and AChE expression were elevated. Discussion This data shows that the 3xTg-AD mouse model can be useful for studying the human sex differences of AD, but only in regards to Ap. Furthermore, in vitro data shows local E2 may be protective for EGR1 and cholinergic functions in AD while suppressing soluble Aβ(1-42) levels. Altogether, this study provides further in vivo and in vitro data supporting the human epidemiological data indicating a higher prevalence of AD in women is related to changes in brain estrogen levels.
Collapse
Affiliation(s)
- Yu-Ting Hu
- Affiliated Mental Health Center and Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xin-Lu Chen
- Department of Neurobiology and Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Ya-Nan Zhang
- Department of Neurobiology and Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
| | - Hugo McGurran
- Netherlands Institute for Neuroscience, The Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Jochem Stormmesand
- Netherlands Institute for Neuroscience, The Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Nicole Breeuwsma
- Netherlands Institute for Neuroscience, The Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Arja Sluiter
- Netherlands Institute for Neuroscience, The Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Juan Zhao
- Netherlands Institute for Neuroscience, The Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
| | - Dick Swaab
- Department of Neurobiology and Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Netherlands Institute for Neuroscience, The Royal Netherlands Academy of Arts and Sciences, Amsterdam, Netherlands
- *Correspondence: Dick Swaab,
| | - Ai-Min Bao
- Department of Neurobiology and Neurology of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC and CAMS Key Laboratory of Medical Neurobiology, MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, China
- Ai-Min Bao,
| |
Collapse
|
4
|
Wang Y, Cui Y, Liu J, Song Q, Cao M, Hou Y, Zhang X, Wang P. Krüppel-like factor 5 accelerates the pathogenesis of Alzheimer’s disease via BACE1-mediated APP processing. Alzheimers Res Ther 2022; 14:103. [PMID: 35883144 PMCID: PMC9316766 DOI: 10.1186/s13195-022-01050-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/13/2022] [Indexed: 11/12/2022]
Abstract
Background The deposition of β-amyloid (Aβ) in the brain plays a major role in the pathogenesis of Alzheimer’s disease (AD). Aβ is generated via amyloid precursor protein (APP) cleavage through the amyloidogenic pathway. In this pathway, β-secretase (BACE1) is the first and rate-limiting enzyme. Its expression increases through an unknown mechanism in patients with AD. Thus, the key regulatory mechanism of BACE1 in the AD process should be revealed to understand the pathogenesis of AD and explore the key treatment targets of AD. Methods Here, APPswe/PS1dE9 (APP/PS1) mice were employed to observe the Krüppel-like factor 5 (KLF5) and BACE1 levels in the serum and brain tissues. HT22 cells were used to explore the relationship between KLF5 and BACE1. Results In this study, KLF5 was found to be a novel transcription factor that positively regulated BACE1 by binding to the BACE1 promoter. The KLF5 levels significantly increased not only in the CSF and serum of patients with AD but also in the brain tissue of APP/PS1 mice. They were closely related to cognitive capacity. KLF5 accelerated APP amyloidogenic metabolism and promoted Aβ synthesis through BACE1. Silencing BACE1 could block the KLF5-induced amyloidogenic process of APP. ML264 ameliorated the cognitive deficits and slowed down APP amyloidogenic cleavage in APP/PS1 mice. Conclusion The findings above suggest that upregulation of KLF5 might be a critical element in AD progression by accelerating BACE1-mediated APP amyloidogenic cleavage. The inhibition of KLF5 or the combined inhibitory effect of KLF5 and the BACE1 promoter might be a potential strategy to prevent AD pathogenesis. Supplementary Information The online version contains supplementary material available at 10.1186/s13195-022-01050-3.
Collapse
|
5
|
An F, Zhao R, Xuan X, Xuan T, Zhang G, Wei C. Calycosin ameliorates advanced glycation end product-induced neurodegenerative changes in cellular and rat models of diabetes-related Alzheimer's disease. Chem Biol Interact 2022; 368:110206. [PMID: 36195188 DOI: 10.1016/j.cbi.2022.110206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/20/2022] [Accepted: 09/28/2022] [Indexed: 11/03/2022]
Abstract
Growing pieces of evidence suggest that Alzheimer's disease (AD) is interlinked with Type 2 diabetes mellitus (DM), which has been described as "type 3 DM". In this study, we investigate the neuronal insult attributable to advanced glycation end products (AGEs) as the models of DM-related AD to understand the effects exerted by calycosin on neurodegenerative changes both in vivo and in vitro studies and also studied the associated molecular mechanisms. The results reported herein revealed that the viability of the PC12 cells induced by AGEs increased when treated with calycosin. It was also observed that the learning and memory abilities of AGE-induced DM-related AD rats improved under these conditions. Analysis of the reported results indicates that calycosin can effectively down-regulate the activity of GSK-3β to result in the reversal of the process of tau hyperphosphorylation, inhibit the expression of RAGE and BACE-1 proteins, resulting in a decrease in the production of β-amyloid and regulate the PGC-1α/TFAM signaling pathway to repair mitochondrial dysfunction. It can be inferred that calycosin can potentially exhibit important therapeutic properties that can be exploited during the treatment of AD, especially DM-related AD.
Collapse
Affiliation(s)
- Fengmao An
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China; Medical College, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China.
| | - Ruyi Zhao
- Department of Medicine and Food, Tongliao Vocational College, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China.
| | - Xinran Xuan
- First Clinical Medical College, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China.
| | - Tianqi Xuan
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China; Medical College, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China.
| | - Guowei Zhang
- College of Nursing, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China; Institute of Dementia, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China.
| | - Chengxi Wei
- Institute of Pharmaceutical Chemistry and Pharmacology, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China; Inner Mongolia Key Laboratory of Mongolian Medicine Pharmacology for Cardio-Cerebral Vascular System, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China; Medical College, Inner Mongolia Minzu University, Tongliao, 028000, Inner Mongolia Autonomous Region, PR China.
| |
Collapse
|
6
|
Rangan P, Lobo F, Parrella E, Rochette N, Morselli M, Stephen TL, Cremonini AL, Tagliafico L, Persia A, Caffa I, Monacelli F, Odetti P, Bonfiglio T, Nencioni A, Pigliautile M, Boccardi V, Mecocci P, Pike CJ, Cohen P, LaDu MJ, Pellegrini M, Xia K, Tran K, Ann B, Chowdhury D, Longo VD. Fasting-mimicking diet cycles reduce neuroinflammation to attenuate cognitive decline in Alzheimer's models. Cell Rep 2022; 40:111417. [PMID: 36170815 PMCID: PMC9648488 DOI: 10.1016/j.celrep.2022.111417] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 03/30/2022] [Accepted: 09/01/2022] [Indexed: 11/22/2022] Open
Abstract
The effects of fasting-mimicking diet (FMD) cycles in reducing many aging and disease risk factors indicate it could affect Alzheimer's disease (AD). Here, we show that FMD cycles reduce cognitive decline and AD pathology in E4FAD and 3xTg AD mouse models, with effects superior to those caused by protein restriction cycles. In 3xTg mice, long-term FMD cycles reduce hippocampal Aβ load and hyperphosphorylated tau, enhance genesis of neural stem cells, decrease microglia number, and reduce expression of neuroinflammatory genes, including superoxide-generating NADPH oxidase (Nox2). 3xTg mice lacking Nox2 or mice treated with the NADPH oxidase inhibitor apocynin also display improved cognition and reduced microglia activation compared with controls. Clinical data indicate that FMD cycles are feasible and generally safe in a small group of AD patients. These results indicate that FMD cycles delay cognitive decline in AD models in part by reducing neuroinflammation and/or superoxide production in the brain.
Collapse
Affiliation(s)
- Priya Rangan
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Fleur Lobo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Edoardo Parrella
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; Department of Molecular and Translational Medicine, University of Brescia, Viale Europa 11, Brescia, BS 25123, Italy
| | - Nicolas Rochette
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA; Department of Ecology and Evolutionary Biology, University of California, Los Angeles, 612 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Marco Morselli
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 610 Charles E. Young Dr. S., Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Terri-Leigh Stephen
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Anna Laura Cremonini
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Luca Tagliafico
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Angelica Persia
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Irene Caffa
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Fiammetta Monacelli
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Patrizio Odetti
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Tommaso Bonfiglio
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy
| | - Alessio Nencioni
- Department of Internal Medicine and Medical Specialties, University of Genoa, Viale Benedetto XV 6, Genova, GE 16132, Italy; IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132 Genoa, Italy
| | - Martina Pigliautile
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Virginia Boccardi
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Patrizia Mecocci
- Santa Maria della Misericordia Hospital, Section of Gerontology and Geriatrics, Department of Medicine and Surgery, University of Perugia, Perugia, Italy
| | - Christian J Pike
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Pinchas Cohen
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; USC Dornsife College of Letters, Arts & Sciences, Department of Biological Sciences, University of Southern California, 3551 Trousdale Pkwy., Los Angeles, CA 90089-0191, USA
| | - Mary Jo LaDu
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, USA
| | - Matteo Pellegrini
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, 610 Charles E. Young Dr. S., Los Angeles, CA 90095, USA; Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, 611 Charles E. Young Dr. E., Los Angeles, CA 90095, USA
| | - Kyle Xia
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Katelynn Tran
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Brandon Ann
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Dolly Chowdhury
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA
| | - Valter D Longo
- Longevity Institute, School of Gerontology, Department of Biological Sciences, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191, USA; Eli and Edythe Broad Center for Regenerative Medicine and Stem Cell Research at USC, Keck School of Medicine, University of Southern California, 1425 San Pablo St., Los Angeles, CA 90033, USA; IFOM FIRC Institute of Molecular Oncology, Via Adamello 16, Milano, MI 20139, Italy.
| |
Collapse
|
7
|
AdipoRon induces AMPK activation and ameliorates Alzheimer's like pathologies and associated cognitive impairment in APP/PS1 mice. Neurobiol Dis 2022; 174:105876. [PMID: 36162737 DOI: 10.1016/j.nbd.2022.105876] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 09/12/2022] [Accepted: 09/21/2022] [Indexed: 11/24/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive devastating neurodegenerative disorder characterized by extracellular amyloid beta (Aβ42) plaque formation, hyperphosphorylation of tau protein leading to intracellular neurofibrillary tangle formation. Recently discovered hallmark features responsible for AD pathogenesis are neuronal insulin resistance, dysregulation in adiponectin and AMPK signaling. The presence of adiponectin and its receptor in the brain with its unique anti-diabetic effects and association with neurodegenerative diseases has raised our interest in exploring orally active small molecule adiponectin receptor agonist, AdipoRon. To date, all the available drugs for the treatment of AD provides symptomatic relief and do not stall the progression of the disease. Indeed, it is becoming increasingly apparent to find appropriate targets. Here, we attempt to shed lights on adiponectin receptor agonist, AdipoRon and its downstream molecular targets in reducing disease pathogenesis and insulin resistance. In brain, AdipoRon induced AMPK activation, increased insulin sensitivity, reduced amyloid beta plaque deposition and improved cognitive impairment. Levels of BACE were also downregulated while LDLR, APOE and neprilysin were upregulated promoting amyloid beta clearance from brain. AdipoRon further reduced the chronic inflammatory marker, GFAP and improved synaptic markers PSD-95 and synaptophysin in APP/PS1 mice. Our in-vitro studies further confirmed the potential role of AdipoRon in improving insulin sensitivity by increasing GLUT 4 translocation, glucose uptake and insulin signaling under hyperinsulinemic condition. Our findings suggest that AdipoRon could be a promising lead in the future treatment strategies in the development of effective AD treatment.
Collapse
|
8
|
Manganese-Induced Toxicity in C. elegans: What Can We Learn from the Transcriptome? Int J Mol Sci 2022; 23:ijms231810748. [PMID: 36142660 PMCID: PMC9502620 DOI: 10.3390/ijms231810748] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/09/2022] [Accepted: 09/13/2022] [Indexed: 11/21/2022] Open
Abstract
Manganese (Mn) is an essential ubiquitous transition metal and, when occupationally or environmentally overexposed, a well-known risk factor for several neurological pathologies. However, the molecular mechanisms underlying Mn-induced neurotoxicity are largely unknown. In this study, addressing RNA-Seq analysis, bioavailability and survival assays, key pathways of transcriptional responses to Mn overexposure were investigated in the model organism Caenorhabditis elegans (C. elegans), providing insights into the Mn-induced cellular stress and damage response. Comparative transcriptome analyses identified a large number of differentially expressed genes (DEGs) in nematodes exposed to MnCl2, and functional annotation suggested oxidative nucleotide damage, unfolded protein response and innate immunity as major damage response pathways. Additionally, a time-dependent increase in the transcriptional response after MnCl2 exposure was identified by means of increased numbers of DEGs, indicating a time-dependent response and activation of the stress responses in Mn neurotoxicity. The data provided here represent a powerful transcriptomic resource in the field of Mn toxicity, and therefore, this study provides a useful basis for further planning of targeted mechanistic studies of Mn-induced neurotoxicity that are urgently needed in the face of increasing industrially caused environmental pollution with Mn.
Collapse
|
9
|
He L, Liu X, Li H, Dong R, Liang R, Wang R. Polyrhachis vicina Roger Alleviates Memory Impairment in a Rat Model of Alzheimer's Disease Through the EGR1/BACE1/APP Axis. ACS Chem Neurosci 2022; 13:1857-1867. [PMID: 35675207 DOI: 10.1021/acschemneuro.1c00193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Memory deficits and loss are the earliest and most prominent features of Alzheimer's disease (AD). This study was aimed to clarify the mechanistic basis of an active fraction of Polyrhachis vicina Roger (AFPR) on the memory abilities of AD rat models, which involves early growth response 1 (EGR1) expression and β-secretase 1 (BACE1)-mediated deposition of amyloid β peptide (Aβ). An AD rat model was developed by Aβ25-35, which was further treated with AFPR alone or in combination with lentiviral EGR1. The Morris water maze test and HE and Fluoro-Jade C staining were adopted to observe the memory behaviors, hippocampus neuron morphology, and Aβ deposition. Aβ25-35-induced SK-N-SH and HT22 neurons were subjected to AFPR for in vitro experiments on neuronal viability and apoptosis. AFPR improved the impaired memory function, preserved the neuron structure, and suppressed Aβ deposition in AD rat models. Further, the expression of APP pathway-related proteins was downregulated by AFPR in both rat and cellular models. Moreover, AFPR inhibited the Aβ25-35-induced neuronal apoptosis. AFPR suppressed the expression of EGR1, downregulated the BACE1 expression via impeding the binding of EGR1 to the BACE1 promoter, and thus blocked the activation of the APP signaling, ultimately protecting neurons. Notably, the aforementioned effects of AFPR were in a concentration-dependent manner; among three doses, 3.65, 15.6, and 30 mg/(kg·d), high-dose AFPR exhibited the most appreciable effects. In conclusion, AFPR inhibited the BACE1 expression by repressing the binding of EGR1 to the promoter of BACE1, thereby suppressing the Aβ deposition and improving the memory function of AD rats.
Collapse
Affiliation(s)
- Luyan He
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Xiaoman Liu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Hualian Li
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Ruifang Dong
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Ruobing Liang
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| | - Ruoxi Wang
- Department of Neurology, Cangzhou Central Hospital, Cangzhou 061000, P.R. China
| |
Collapse
|
10
|
EGR1 Enhances Lymphangiogenesis via SOX18-Mediated Activation of JAK2/STAT3 Pathway. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6448724. [PMID: 35190753 PMCID: PMC8858051 DOI: 10.1155/2022/6448724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/27/2021] [Accepted: 12/29/2021] [Indexed: 11/18/2022]
Abstract
Background. Lymphangiogenesis is a process involved in the pathogenesis of many diseases. Identifying key molecules and pathway targeting this process is critical for lymphatic regeneration-associated disorders. EGR1 is a transcription factor, but its function in lymphangiogenesis is not yet known. This study is aimed at exploring the functional activity and molecular mechanism of EGR1 implicated in lymphangiogenesis. Methods. The CCK-8 method, transwell migration assay, and tube formation assay were used to detect the cell viability, motility, and tube formation of HDLEC cells, respectively. The luciferase reporter assay was applied to detect the impact of EGR1 on SOX18 promoter activity. CHIP assay was used to analyze the direct binding of EGR1 to the SOX18 promoter. qRT-PCR and Western blot analysis were performed to investigate molecules and pathway involved in lymphangiogenesis. Results. The EGR1 ectopic expression markedly increased the cell growth, mobility, tube formation, and the expression of lymphangiogenesis-associated markers (LYVE-1 and PROX1) in HDLEC cells. EGR1 interacted with the SXO18 gene promoter and transcriptionally regulated the SXO18 expression in HDLEC cells. Silencing of SOX18 abrogated the promotional activities of EGR1 on the cell viability, mobility, tube formation, and LYVE-1/PROX1 expression in HDLEC cells. SOX18 overexpression activated JAK/STAT signaling, which resulted in an increase in lymphangiogenesis in HDLEC cells. Conclusions. ERG1 can promote lymphangiogenesis, which is mediated by activating the SOX18/JAK/STAT3 cascade. ERG1 may serve as a promising target for the therapy of lymphatic vessel-related disorders.
Collapse
|
11
|
Pang Z, Xu Y, Zhu Q. Early Growth Response 1 Suppresses Macrophage Phagocytosis by Inhibiting NRF2 Activation Through Upregulation of Autophagy During Pseudomonas aeruginosa Infection. Front Cell Infect Microbiol 2022; 11:773665. [PMID: 35096638 PMCID: PMC8790152 DOI: 10.3389/fcimb.2021.773665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 12/22/2021] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that causes life-threatening infections in cystic fibrosis patients and immunocompromised individuals. A tightly regulated immune response possessed by healthy individuals can effectively control P. aeruginosa infections, whereas the patients with dysregulated immune response are susceptible to this bacterial pathogen. Early growth response 1 (Egr-1) is a zinc-finger transcription factor involved in regulation of various cellular functions, including immune responses. We previously identified that Egr-1 was deleterious to host in a mouse model of acute P. aeruginosa pneumonia by promoting systemic inflammation and impairing bacterial clearance in lung, which associated with reduced phagocytosis and bactericidal ability of leucocytes, including macrophages and neutrophils. However, the molecular mechanisms underlying the Egr-1-suppressed phagocytosis of P. aeruginosa are incompletely understood. Herein, we investigated whether the Egr-1-regulated autophagy play a role in macrophage phagocytosis during P. aeruginosa infection by overexpression or knockdown of Egr-1. We found that overexpression of Egr-1 inhibited the phagocytic activity of macrophages, and the autophagy activator rapamycin and inhibitor chloroquine could reverse the effects of Egr-1 knockdown and Egr-1 overexpression on phagocytosis of P. aeruginosa, respectively. Furthermore, the Egr-1-overexpressing macrophages displayed upregulated expression of autophagy-related proteins LC3A, LC3B and Atg5, and decreased levels of p62 in macrophages. Further studies revealed that the macrophages with Egr-1 knockdown displayed enhanced activation of transcription factor NRF2 and expression of scavenger receptors MACRO and MSR1. Altogether, these findings suggest that Egr-1 suppresses the phagocytosis of P. aeruginosa by macrophages through upregulation of autophagy and inhibition of NRF2 signaling.
Collapse
Affiliation(s)
- Zheng Pang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Xu
- College of Traditional Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qingjun Zhu
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China.,Key Laboratory of Traditional Chinese Medicine Classical Theory, Ministry of Education, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
12
|
Comparative Transcriptome Analysis in Monocyte-Derived Macrophages of Asymptomatic GBA Mutation Carriers and Patients with GBA-Associated Parkinson's Disease. Genes (Basel) 2021; 12:genes12101545. [PMID: 34680941 PMCID: PMC8535749 DOI: 10.3390/genes12101545] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/31/2022] Open
Abstract
Mutations of the GBA gene, encoding for lysosomal enzyme glucocerebrosidase (GCase), are the greatest genetic risk factor for Parkinson’s disease (PD) with frequency between 5% and 20% across the world. N370S and L444P are the two most common mutations in the GBA gene. PD carriers of severe mutation L444P in the GBA gene is characterized by the earlier age at onset compared to N370S. Not every carrier of GBA mutations develop PD during one’s lifetime. In the current study we aimed to find common gene expression signatures in PD associated with mutation in the GBA gene (GBA-PD) using RNA-seq. We compared transcriptome of monocyte-derived macrophages of 5 patients with GBA-PD (4 L444P/N, 1 N370S/N) and 4 asymptomatic GBA mutation carriers (GBA-carriers) (3 L444P/N, 1 N370S/N) and 4 controls. We also conducted comparative transcriptome analysis for L444P/N only GBA-PD patients and GBA-carriers. Revealed deregulated genes in GBA-PD independently of GBA mutations (L444P or N370S) were involved in immune response, neuronal function. We found upregulated pathway associated with zinc metabolism in L444P/N GBA-PD patients. The potential important role of DUSP1 in the pathogenesis of GBA-PD was suggested.
Collapse
|
13
|
Scarpa JR, Jiang P, Gao VD, Vitaterna MH, Turek FW, Kasarskis A. NREM delta power and AD-relevant tauopathy are associated with shared cortical gene networks. Sci Rep 2021; 11:7797. [PMID: 33833255 PMCID: PMC8032807 DOI: 10.1038/s41598-021-86255-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 07/27/2020] [Indexed: 02/01/2023] Open
Abstract
Reduced NREM sleep in humans is associated with AD neuropathology. Recent work has demonstrated a reduction in NREM sleep in preclinical AD, pointing to its potential utility as an early marker of dementia. We test the hypothesis that reduced NREM delta power and increased tauopathy are associated with shared underlying cortical molecular networks in preclinical AD. We integrate multi-omics data from two extensive public resources, a human Alzheimer's disease cohort from the Mount Sinai Brain Bank (N = 125) reflecting AD progression and a (C57BL/6J × 129S1/SvImJ) F2 mouse population in which NREM delta power was measured (N = 98). Two cortical gene networks, including a CLOCK-dependent circadian network, are associated with NREM delta power and AD tauopathy progression. These networks were validated in independent mouse and human cohorts. Identifying gene networks related to preclinical AD elucidate possible mechanisms associated with the early disease phase and potential targets to alter the disease course.
Collapse
Affiliation(s)
- Joseph R Scarpa
- Department of Anesthesiology, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Peng Jiang
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Vance D Gao
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Martha H Vitaterna
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Fred W Turek
- Center for Sleep and Circadian Biology, Department of Neurobiology, Northwestern University, Evanston, IL, 60208, USA
| | - Andrew Kasarskis
- Icahn Institute for Genomics and Multiscale Biology, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Population Health Science and Policy, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
14
|
He K, Zhang J, Liu J, Cui Y, Liu LG, Ye S, Ban Q, Pan R, Liu D. Functional genomics study of protein inhibitor of activated STAT1 in mouse hippocampal neuronal cells revealed by RNA sequencing. Aging (Albany NY) 2021; 13:9011-9027. [PMID: 33759814 PMCID: PMC8034905 DOI: 10.18632/aging.202749] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
Protein inhibitor of activated STAT1 (PIAS1), a small ubiquitin-like modifier (SUMO) E3 ligase, was considered to be an inhibitor of STAT1 by inhibiting the DNA-binding activity of STAT1 and blocking STAT1-mediated gene transcription in response to cytokine stimulation. PIAS1 has been determined to be involved in modulating several biological processes such as cell proliferation, DNA damage responses, and inflammatory responses, both in vivo and in vitro. However, the role played by PIAS1 in regulating neurodegenerative diseases, including Alzheimer’s disease (AD), has not been determined. In our study, significantly different expression levels of PIAS1 between normal controls and AD patients were detected in four regions of the human brain. Based on a functional analysis of Pias1 in undifferentiated mouse hippocampal neuronal HT-22 cells, we observed that the expression levels of several AD marker genes could be inhibited by Pias1 overexpression. Moreover, the proliferation ability of HT-22 cells could be promoted by the overexpression of Pias1. Furthermore, we performed RNA sequencing (RNA-seq) to evaluate and quantify the gene expression profiles in response to Pias1 overexpression in HT-22 cells. As a result, 285 significantly dysregulated genes, including 79 upregulated genes and 206 downregulated genes, were identified by the comparison of Pias1/+ cells with WT cells. Among these genes, five overlapping genes, including early growth response 1 (Egr1), early growth response 2 (Egr2), early growth response 3 (Egr3), FBJ osteosarcoma oncogene (Fos) and fos-like antigen 1 (Fosl1), were identified by comparison of the transcription factor binding site (TFBS) prediction results for STAT1, whose expression was evaluated by qPCR. Three cell cycle inhibitors, p53, p18 and p21, were significantly downregulated with the overexpression of Pias1. Analysis of functional enrichment and expression levels showed that basic region leucine zipper domain-containing transcription factors including zinc finger C2H2 (zf-C2H2), homeobox and basic/helix-loop-helix (bHLH) in several signaling pathways were significantly involved in PIAS1 regulation in HT-22 cells. A reconstructed regulatory network under PIAS1 overexpression demonstrated that there were 43 related proteins, notably Nr3c2, that directly interacted with PIAS1.
Collapse
Affiliation(s)
- Kan He
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China.,Department of Biostatistics, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Jian Zhang
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Justin Liu
- Department of Statistics, University of California, Riverside, CA 92521, USA
| | - Yandi Cui
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | | | - Shoudong Ye
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China.,Department of Biostatistics, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Qian Ban
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China.,Department of Biostatistics, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Ruolan Pan
- Center for Stem Cell and Translational Medicine, School of Life Sciences, Anhui University, Hefei 230601, Anhui, China
| | - Dahai Liu
- Foshan Stomatology Hospital, School of Medicine, Foshan University, Foshan 528000, Guangdong, China
| |
Collapse
|
15
|
Yee A, Tsui NBY, Kwan RYC, Leung AYM, Lai CKY, Chung T, Lau JYN, Fok M, Dai DLK, Lau LT. Apolipoprotein E Gene Revisited: Contribution of Rare Variants to Alzheimer's Disease Susceptibility in Southern Chinese. Curr Alzheimer Res 2021; 18:67-79. [PMID: 33761857 DOI: 10.2174/1567205018666210324111401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 12/28/2020] [Accepted: 02/16/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND APOE ε4 is the best-known risk factor for late-onset alzheimer's disease (AD). Population studies have demonstrated a relatively low prevalence of APOE ε4 among Chinese population, implying additional risk factors that are Chinese-specific may exist. Apart from - alleles, genetic variation profile along the full-length APOE has rarely been investigated. OBJECTIVE In this study, we filled this gap by comprehensively determining all genetic variations in APOE and investigated their potential associations with late-onset AD and mild cognitive impairment (MCI) in southern Chinese. METHODS Two hundred and fifty-seven southern Chinese participants were recruited, of whom 69 were AD patients, 83 had MCI, and 105 were normal controls. Full-length APOE from promoter to 3'UTR regions were sequenced. Genetic variants were identified and compared among the three groups. RESULTS While APOE ε4 was more significantly found in AD patients, the prevalence of APOE ε4 in southern Chinese AD patients was the lowest when compared to other areas of China and nearby regions, as well as other countries worldwide. We further identified 13 rare non-singleton variants in APOE. Significantly more AD patients carried any of the rare non-singleton variants than MCI and normal subjects. Such difference was observed in the non-carriers of ε4-allele only. Among the identified rare variants, the potential functional impact was predicted for rs532314089, rs553874843, rs533904656 and rs370594287. CONCLUSION Our study suggests an ethnic difference in genetic risk composition of AD in southern Chinese. Rare variants on APOE are a potential candidate for AD risk stratification biomarker in addition to APOE-ε4.
Collapse
Affiliation(s)
- Anita Yee
- Avalon Genomics (Hong Kong) Limited, Shatin,Hong Kong
| | | | - Rick Y C Kwan
- Centre for Gerontological Nursing, School of Nursing, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| | - Angela Y M Leung
- Centre for Gerontological Nursing, School of Nursing, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| | - Claudia K Y Lai
- Centre for Gerontological Nursing, School of Nursing, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| | - Teresa Chung
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| | | | - Manson Fok
- Faculty of Health Sciences, Macau University of Science and Technology, Taipa, Macau,Hong Kong
| | - David L K Dai
- Hong Kong Alzheimer's Disease Association, Wang Tau Hom,Hong Kong
| | - Lok-Ting Lau
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom,Hong Kong
| |
Collapse
|
16
|
Dimeric Transmembrane Orientations of APP/C99 Regulate γ-Secretase Processing Line Impacting Signaling and Oligomerization. iScience 2020; 23:101887. [PMID: 33367225 PMCID: PMC7749410 DOI: 10.1016/j.isci.2020.101887] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/21/2020] [Accepted: 11/25/2020] [Indexed: 12/22/2022] Open
Abstract
Amyloid precursor protein (APP) cleavage by the β-secretase produces the C99 transmembrane (TM) protein, which contains three dimerization-inducing Gly-x-x-x-Gly motifs. We demonstrate that dimeric C99 TM orientations regulate the precise cleavage lines by γ-secretase. Of all possible dimeric orientations imposed by a coiled-coil to the C99 TM domain, the dimer containing the 33Gly-x-x-x-Gly37 motif in the interface promoted the Aβ42 processing line and APP intracellular domain-dependent gene transcription, including the induction of BACE1 mRNA, enhancing amyloidogenic processing and signaling. Another orientation exhibiting the 25Gly-x-x-x-Gly29 motif in the interface favored processing to Aβ43/40. It induced significantly less gene transcription, while promoting formation of SDS-resistant "Aβ-like" oligomers, reminiscent of Aβ peptide oligomers. These required both Val24 of a pro-β motif and the 25Gly-x-x-x-Gly29 interface. Thus, crossing angles imposed by precise dimeric orientations control γ-secretase initial cleavage at Aβ48 or Aβ49, linking the former to enhanced signaling and Aβ42 production.
Collapse
|
17
|
Xiang Y, Peng J, Liu Q, Guo C. The association between CD69 and EGR1 levels, and CHD patients without reflow after PCI. Exp Ther Med 2019; 17:3913-3920. [PMID: 30988776 PMCID: PMC6447924 DOI: 10.3892/etm.2019.7411] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 01/30/2019] [Indexed: 12/21/2022] Open
Abstract
Association between CD69 and EGR1 levels and coronary heart disease (CHD) patients without reflow after percutaneous coronary intervention (PCI) was investigated. Data of 156 patients undergoing PCI from September 2014 to December 2016 in People's Hospital of Hunan Province were analyzed, and they were divided into 72 cases in reflow group (group A) and 84 cases in no-reflow group (group B) according to the patient's postoperative blood flow. Another 70 volunteers undergoing normal physical examination in the same period were selected as control group (group C). The venous blood was extracted from patients before operation, at 5 min, 2 h and 6 months after operation, the serum CD69 and EGR1 levels were detected with flow cytometry and RT-qPCR methods, and association between CD69 and EGR1 levels and postoperative blood flow recovery was analyzed. CD69 expression before and 2 h after operation in group A was significant (P<0.05), and a difference in patients at 6 months after operation between group A, B and group C (P<0.05). The CD69 expression in group A was significantly lower than that in group B (P<0.05), but in group B was significantly higher than that in group C (P<0.05). The EGR1 expression in group A was significantly higher than that in group B (P<0.05), but in group B was significantly lower than that in group C (P<0.05). Multivariate logistic regression analysis revealed that CD69 (OR=6.424, P=0.025) and EGR1 (OR=3.684, P=0.013) were independent risk factors for patients without reflow after undergoing PCI. After undergoing PCI in CHD patients, if there was an increase in CD69 level and a significant decrease in EGR1 level in the early postoperative period, the patient may be suspected of having no-reflow and checked in time to improve the patient's therapeutic effect.
Collapse
Affiliation(s)
- Yi Xiang
- Department of Geriatrics, People's Hospital of Hunan Province, Changsha, Hunan 410006, P.R. China
| | - Jianqiao Peng
- Department of Clinical Laboratory, People's Hospital of Hunan Province, Changsha, Hunan 410006, P.R. China
| | - Qiong Liu
- Department of Clinical Laboratory, People's Hospital of Hunan Province, Changsha, Hunan 410006, P.R. China
| | - Chun Guo
- Department of Pediatrics, People's Hospital of Hunan Province, Changsha, Hunan 410006, P.R. China
| |
Collapse
|
18
|
Zhao X, Wang X, Su G, Sun Q, Fu J, Zhang H, Teng J. The effect of early growth response 1 on levels of Amyloid-β 40 peptide in U87MG cells. J Cell Biochem 2018; 120:3514-3519. [PMID: 30548663 DOI: 10.1002/jcb.27627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/14/2018] [Indexed: 11/11/2022]
Abstract
A recent study has shown that early growth response 1 (EGR1) plays a critical role in the β-amyloid cascade and tau hypotheses. In addition, evidence has suggested that EGR1 can regulate levels of amyloid-beta peptides, key molecules in the pathogenesis of Alzheimer's disease (AD). However, whether EGR1 is a deleterious or protective factor in the AD is still controversial. In this present study, we constructed an overexpression plasmid, CMV-EGFP-EGR1-Kanamycin, and transfected it into U87MG cells to investigate the effects of EGR1 expression on amyloid-β (1-40) peptide (Aβ40) levels. U87MG cells transfected by CMV-EGFP-EGR1-Kanamycin and CMV-EGFP-Kanamycin were assigned, respectively, to experimental and control groups. Fluorescence microscopy was used to observe transfection efficiencies of the plasmids after 6 hours. EGR1 messenger RNA levels were measured by quantitative reverse transcription polymerase chain reaction. Aβ40 secretion was analyzed by enzyme-linked immunosorbent assay. Expression of the amyloid precursor protein, beta-secretase enzyme, and presenilin 1 proteins were analyzed by Western blot analysis. The results showed that EGR1 overexpression increased Aβ40 secretion in vitro, possibly through increasing BACE1 expression. Based on these results, EGR1 might be a promising therapeutic target for the AD.
Collapse
Affiliation(s)
- Xinyu Zhao
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiaojie Wang
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Gang Su
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qi Sun
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jitong Fu
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huili Zhang
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junfang Teng
- Department of Neurology, The Frist Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
19
|
Lesuis SL, Hoeijmakers L, Korosi A, de Rooij SR, Swaab DF, Kessels HW, Lucassen PJ, Krugers HJ. Vulnerability and resilience to Alzheimer's disease: early life conditions modulate neuropathology and determine cognitive reserve. Alzheimers Res Ther 2018; 10:95. [PMID: 30227888 PMCID: PMC6145191 DOI: 10.1186/s13195-018-0422-7] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 08/15/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND Alzheimer's disease (AD) is a progressive neurodegenerative disorder with a high prevalence among the elderly and a huge personal and societal impact. Recent epidemiological studies have indicated that the incidence and age of onset of sporadic AD can be modified by lifestyle factors such as education, exercise, and (early) stress exposure. Early life adversity is known to promote cognitive decline at a later age and to accelerate aging, which are both primary risk factors for AD. In rodent models, exposure to 'negative' or 'positive' early life experiences was recently found to modulate various measures of AD neuropathology, such as amyloid-beta levels and cognition at later ages. Although there is emerging interest in understanding whether experiences during early postnatal life also modulate AD risk in humans, the mechanisms and possible substrates underlying these long-lasting effects remain elusive. METHODS We review literature and discuss the role of early life experiences in determining later age and AD-related processes from a brain and cognitive 'reserve' perspective. We focus on rodent studies and the identification of possible early determinants of later AD vulnerability or resilience in relation to early life adversity/enrichment. RESULTS Potential substrates and mediators of early life experiences that may influence the development of AD pathology and cognitive decline are: programming of the hypothalamic-pituitary-adrenal axis, priming of the neuroinflammatory response, dendritic and synaptic complexity and function, overall brain plasticity, and proteins such as early growth response protein 1 (EGR1), activity regulated cytoskeleton-associated protein (Arc), and repressor element-1 silencing transcription factor (REST). CONCLUSIONS We conclude from these rodent studies that the early postnatal period is an important and sensitive phase that influences the vulnerability to develop AD pathology. Yet translational studies are required to investigate whether early life experiences also modify AD development in human studies, and whether similar molecular mediators can be identified in the sensitivity to develop AD in humans.
Collapse
Affiliation(s)
- Sylvie L. Lesuis
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Lianne Hoeijmakers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Aniko Korosi
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Susanne R. de Rooij
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
- Department of Clinical Epidemiology, Biostatistics & Bio informatics, Academic Medical Centre, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands
| | - Dick F. Swaab
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
| | - Helmut W. Kessels
- The Netherlands Institute for Neuroscience, an Institute of the Royal Netherlands Academy of Arts and Sciences, KNAW, Meibergdreef 47, 1105 BA Amsterdam, The Netherlands
- Department of Cellular and Computational Neuroscience, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Paul J. Lucassen
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| | - Harm J. Krugers
- Brain Plasticity Group, SILS-CNS, University of Amsterdam, Science Park 904, 1098 XH Amsterdam, The Netherlands
| |
Collapse
|
20
|
Robbins JP, Perfect L, Ribe EM, Maresca M, Dangla-Valls A, Foster EM, Killick R, Nowosiad P, Reid MJ, Polit LD, Nevado AJ, Ebner D, Bohlooly-Y M, Buckley N, Pangalos MN, Price J, Lovestone S. Clusterin Is Required for β-Amyloid Toxicity in Human iPSC-Derived Neurons. Front Neurosci 2018; 12:504. [PMID: 30090055 PMCID: PMC6068261 DOI: 10.3389/fnins.2018.00504] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 07/04/2018] [Indexed: 02/01/2023] Open
Abstract
Our understanding of the molecular processes underlying Alzheimer's disease (AD) is still limited, hindering the development of effective treatments, and highlighting the need for human-specific models. Advances in identifying components of the amyloid cascade are progressing, including the role of the protein clusterin in mediating β-amyloid (Aβ) toxicity. Mutations in the clusterin gene (CLU), a major genetic AD risk factor, are known to have important roles in Aβ processing. Here we investigate how CLU mediates Aβ-driven neurodegeneration in human induced pluripotent stem cell (iPSC)-derived neurons. We generated a novel CLU-knockout iPSC line by CRISPR/Cas9-mediated gene editing to investigate Aβ-mediated neurodegeneration in cortical neurons differentiated from wild type and CLU knockout iPSCs. We measured response to Aβ using an imaging assay and measured changes in gene expression using qPCR and RNA sequencing. In wild type neurons imaging indicated that neuronal processes degenerate following treatment with Aβ25-35 peptides and Aβ1-42 oligomers, in a dose dependent manner, and that intracellular levels of clusterin are increased following Aβ treatment. However, in CLU knockout neurons Aβ exposure did not affect neurite length, suggesting that clusterin is an important component of the amyloid cascade. Transcriptomic data were analyzed to elucidate the pathways responsible for the altered response to Aβ in neurons with the CLU deletion. Four of the five genes previously identified as downstream to Aβ and Dickkopf-1 (DKK1) proteins in an Aβ-driven neurotoxic pathway in rodent cells were also dysregulated in human neurons with the CLU deletion. AD and lysosome pathways were the most significantly dysregulated pathways in the CLU knockout neurons, and pathways relating to cytoskeletal processes were most dysregulated in Aβ treated neurons. The absence of neurodegeneration in the CLU knockout neurons in response to Aβ compared to the wild type neurons supports the role of clusterin in Aβ-mediated AD pathogenesis.
Collapse
Affiliation(s)
| | - Leo Perfect
- Department of Basic and Clinical Neuroscience, King's College London, London, United Kingdom
| | - Elena M Ribe
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Marcello Maresca
- Translational Genomics, Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | | | | | - Richard Killick
- Department of Old Age Psychiatry, King's College London, London, United Kingdom
| | - Paulina Nowosiad
- Department of Basic and Clinical Neuroscience, King's College London, London, United Kingdom
| | - Matthew J Reid
- Department of Basic and Clinical Neuroscience, King's College London, London, United Kingdom
| | - Lucia Dutan Polit
- Department of Basic and Clinical Neuroscience, King's College London, London, United Kingdom
| | - Alejo J Nevado
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Daniel Ebner
- Nuffield Department of Medicine, Target Discovery Institute, University of Oxford, Oxford, United Kingdom
| | - Mohammad Bohlooly-Y
- Translational Genomics, Discovery Sciences, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Noel Buckley
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Menelas N Pangalos
- Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Cambridge, United Kingdom
| | - Jack Price
- Department of Basic and Clinical Neuroscience, King's College London, London, United Kingdom
| | - Simon Lovestone
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
21
|
Cystatin C promotes tau protein phosphorylation and causes microtubule instability by inhibiting intracellular turnover of GSK3β in neurons. Mol Cell Neurosci 2018; 89:1-8. [PMID: 29577984 DOI: 10.1016/j.mcn.2018.03.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 03/16/2018] [Accepted: 03/19/2018] [Indexed: 12/20/2022] Open
Abstract
In Alzheimer's disease (AD) tau protein hyperphosphorylation causes neurofibrillary tangle formation, microtubule instability and neurodegeneration. Determining the mechanism of tau hyperphosphorylation will provide a better understanding of AD pathology. Cystatin C (CysC) is a risk factor for late-onset AD and its level is upregulated in the brains of AD patients. The role of CysC is AD pathogenesis is not known. In this study, we found that CysC level is upregulated in 3xTg-AD mouse brain. We demonstrate that CysC does not affect cellular Aβ production. However, when overexpressed in neuron (NGF-differentiated PC12 cells), CysC inhibits turnover of GSK3β, promotes GSK3β-catalyzed tau phosphorylation at Ser396/404 and causes microtubule instability. Our data provide a novel insight into the role of CysC in AD pathogenesis.
Collapse
|
22
|
MacDonald R, Barbat-Artigas S, Cho C, Peng H, Shang J, Moustaine A, Carbonetto S, Robitaille R, Chalifour LE, Paudel H. A Novel Egr-1-Agrin Pathway and Potential Implications for Regulation of Synaptic Physiology and Homeostasis at the Neuromuscular Junction. Front Aging Neurosci 2017; 9:258. [PMID: 28824419 PMCID: PMC5541023 DOI: 10.3389/fnagi.2017.00258] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/19/2017] [Indexed: 12/11/2022] Open
Abstract
Synaptic transmission requires intricate coordination of the components involved in processing of incoming signals, formation and stabilization of synaptic machinery, neurotransmission and in all related signaling pathways. Changes to any of these components cause synaptic imbalance and disruption of neuronal circuitry. Extensive studies at the neuromuscular junction (NMJ) have greatly aided in the current understanding of synapses and served to elucidate the underlying physiology as well as associated adaptive and homeostatic processes. The heparan sulfate proteoglycan agrin is a vital component of the NMJ, mediating synaptic formation and maintenance in both brain and muscle, but very little is known about direct control of its expression. Here, we investigated the relationship between agrin and transcription factor early growth response-1 (Egr-1), as Egr-1 regulates the expression of many genes involved in synaptic homeostasis and plasticity. Using chromatin immunoprecipitation (ChIP), cell culture with cell lines derived from brain and muscle, and animal models, we show that Egr-1 binds to the AGRN gene locus and suppresses its expression. When compared with wild type (WT), mice deficient in Egr-1 (Egr-1−/−) display a marked increase in AGRN mRNA and agrin full-length and cleavage fragment protein levels, including the 22 kDa, C-terminal fragment in brain and muscle tissue homogenate. Because agrin is a crucial component of the NMJ, we explored possible physiological implications of the Egr-1-agrin relationship. In the diaphragm, Egr-1−/− mice display increased NMJ motor endplate density, individual area and area of innervation. In addition to increased density, soleus NMJs also display an increase in fragmented and faint endplates in Egr-1−/− vs. WT mice. Moreover, the soleus NMJ electrophysiology of Egr-1−/− mice revealed increased quantal content and motor testing showed decreased movement and limb muscle strength compared with WT. This study provides evidence for the potential involvement of a novel Egr-1-agrin pathway in synaptic homeostatic and compensatory mechanisms at the NMJ. Synaptic homeostasis is greatly affected by the process of aging. These and other data suggest that changes in Egr-1 expression may directly or indirectly promote age-related pathologies.
Collapse
Affiliation(s)
- Ryen MacDonald
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada
| | | | - Chulmin Cho
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada
| | - Huashan Peng
- Center for Research in NeuroscienceMontreal, QC, Canada
| | - Jijun Shang
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada
| | - Ayman Moustaine
- Département de neurosciences, Université de MontréalMontreal, QC, Canada
| | - Salvatore Carbonetto
- Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada.,Center for Research in NeuroscienceMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| | - Richard Robitaille
- Département de neurosciences, Université de MontréalMontreal, QC, Canada
| | - Lorraine E Chalifour
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| | - Hemant Paudel
- Lady Davis Institute for Medical Research, Jewish General HospitalMontreal, QC, Canada.,Integrated Program in Neuroscience, McGill UniversityMontreal, QC, Canada.,Department of Medicine, McGill UniversityMontreal, QC, Canada
| |
Collapse
|
23
|
Qin X, Wang Y, Paudel HK. Inhibition of Early Growth Response 1 in the Hippocampus Alleviates Neuropathology and Improves Cognition in an Alzheimer Model with Plaques and Tangles. THE AMERICAN JOURNAL OF PATHOLOGY 2017. [PMID: 28641077 DOI: 10.1016/j.ajpath.2017.04.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
A sporadic form of Alzheimer disease (AD) and vascular dementia share many risk factors, and their pathogenic mechanisms are suggested to be related. Transcription factor early growth response 1 (Egr-1) regulates various vascular pathologies and is up-regulated in both AD brains and AD mouse models; however, its role in AD pathogenesis is unclear. Herein, we report that silencing of Egr-1 in the hippocampus by shRNA reduces tau phosphorylation, lowers amyloid-β (Aβ) pathology, and improves cognition in the 3xTg-AD mouse model. Egr-1 silencing does not affect levels of cyclin-dependent protein kinase 5 (Cdk5), glycogen synthase kinase 3β, protein phosphatase 1, or protein phosphatase 2A, but reduces p35 subunit of Cdk5. Egr-1 silencing also reduces levels of β-secretase 1 (BACE-1) and BACE-1-cleaved amyloid precursor protein (APP) metabolites (secreted APPβ, C99, Aβ40, and Aβ42) but has no effect on presenilin 1 and presenilin 2. In hippocampal primary neurons, Egr-1 binds to BACE-1 and p35 promoters, enhances tau phosphorylation, activates Cdk5 and BACE-1, and accelerates amyloidogenic APP processing. Blocking Cdk5 action blocks Egr-1-induced tau phosphorylation but has no effect on BACE-1 activation and amyloidogenic APP processing. Blocking BACE-1 action, on the other hand, blocks Egr-1-induced amyloidogenic APP processing but does not affect tau phosphorylation. Egr-1 regulates tau phosphorylation and Aβ synthesis in the brain by respectively controlling activities of Cdk5 and BACE-1, suggesting that Egr-1 is a potential therapeutic candidate for the treatment of AD.
Collapse
Affiliation(s)
- Xike Qin
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Yunling Wang
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada
| | - Hemant K Paudel
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, Quebec, Canada.
| |
Collapse
|
24
|
Cho C, MacDonald R, Shang J, Cho MJ, Chalifour LE, Paudel HK. Early growth response-1-mediated down-regulation of drebrin correlates with loss of dendritic spines. J Neurochem 2017; 142:56-73. [DOI: 10.1111/jnc.14031] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Revised: 02/28/2017] [Accepted: 03/22/2017] [Indexed: 12/16/2022]
Affiliation(s)
- Chulmin Cho
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Ryen MacDonald
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Jijun Shang
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Moon Jeong Cho
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
| | - Lorraine E. Chalifour
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
- Department of Medicine; McGill University; Montreal Quebec Canada
| | - Hemant K. Paudel
- Department of Neurology and Neurosurgery; McGill University; Montreal Quebec Canada
- Lady Davis Institute for Medical Research; Jewish General Hospital; Montreal Quebec Canada
- Department of Medicine; McGill University; Montreal Quebec Canada
| |
Collapse
|
25
|
Wang Y, Qin X, Paudel HK. Amyloid β peptide promotes lysosomal degradation of clusterin via sortilin in hippocampal primary neurons. Neurobiol Dis 2017; 103:78-88. [PMID: 28396259 DOI: 10.1016/j.nbd.2017.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 03/29/2017] [Accepted: 04/05/2017] [Indexed: 12/11/2022] Open
Abstract
Progressive accumulation of amyloid-β peptide (Aβ) in the brain is implicated as the central event in the development of Alzheimer's disease (AD). It is thought that extracellular Aβ triggers toxic signals leading to neurodegeneration. The events downstream of Aβ however are not entirely clear. Clusterin (Apo J) is one of the major risk factors for sporadic form of AD. Clusterin binds to Aβ and prevents Aβ aggregation. In addition, clusterin promotes Aβ degradation and accelerates Aβ clearance from the brain. Clusterin thus protects neurons from Aβ and loss of clusterin level in the brain is implicated as promoting AD pathology. In this study, we found that the level of clusterin protein but not mRNA is reduced in the brains of 3xTg-AD mice. When rat hippocampal primary neurons were treated with Aβ1-42, level of clusterin protein but not mRNA was downregulated. Aβ1-42-induced downregulation of clusterin was blocked by lysosome inhibitors bafilomycin A1 and ammonium chloride. In neurons, Aβ1-42 induced expression of sortilin, a lysosomal sorting protein that targets proteins to lysosome for degradation. In BE(2) M17 human neuroblastoma cells, clusterin bound to sortilin and when sortilin expression was silenced, Aβ1-42-induced clusterin downregulation was almost completely blocked. Our data demonstrate that in neurons, Aβ1-42 promotes lysosomal degradation of clusterin by inducing expression of sortilin and provide a novel mechanism by which Aβ promotes AD pathogenesis.
Collapse
Affiliation(s)
- Yunling Wang
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec H4H 1R3, Canada
| | - Xike Qin
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec H4H 1R3, Canada
| | - Hemant K Paudel
- Lady Davis Institute for Medical Research, Jewish General Hospital, Montreal, Quebec H4H 1R3, Canada; Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H4H 1R3, Canada.
| |
Collapse
|